The Dark Side of the COVID-19 Treatments on Mycobacterium tuberculosis Infection 

Flavio De Maio1,2, Delia Mercedes Bianco2 and Giovanni Delogu2,3.

Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy.
2 Dipartimento di Scienze biotecnologiche di base, cliniche intensivologiche e perioperatorie – Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy.
3 Mater Olbia Hospital, Olbia, Italy.

Correspondence to: Flavio De Maio. Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy. E-mail: flavio.demaio@unicatt.it

ublished: March 1, 2022
Received: September 20, 2021
Accepted: February 11, 2022
Mediterr J Hematol Infect Dis 2022, 14(1): e2022021 DOI 10.4084/MJHID.2022.021

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Since the emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) at the end of 2019, a number of medications have been used to treat the infection and the related Coronavirus disease – 19 (COVID-19).
Some of the administered drugs were tested or used in practice only on the basis of biological plausibility; a promising strategy was to target the host immune response, with host directed therapies (HDTs), to reduce systemic hyperinflammation and hypercytokinemia responsible for additional tissue damage.
We summarize the treatments against SARS-CoV-2 and underline their possible effects on Mycobacterium tuberculosis (Mtb) infection. Both SARS-CoV-2 and Mtb respiratory infections impair the host’s immune response. Furthermore, little research has been conducted on the impact of medicaments used to counteract COVID-19 disease in patients with Latent Tuberculosis Infection (LTBI). A number of these drugs may modulate host immune response by modifying LTBI dynamic equilibrium, favoring either the host or the bacteria.



Introduction

COVID-19 pandemic has shown a significant disruptive impact on Tuberculosis (TB) services, with negative effects on prompt diagnosis, treatment and immunization.[1,2] Pressure on laboratories and pharmaceutical industries led to the readaptation of many TB labs to detect SARS-CoV-2 as well as Bacillus Calmette-Guérin (BCG) shortages and consequent decrease of newborn vaccinations.[3] Estimates indicate a 25% drop in the global BCG coverage and an increase in pediatric deaths ranging from 0.5% to 17%.[4]
In several countries, reports suggest a decline in case notification in the last few months due to massive cancellation of routine health services in many
settings.[5-7] Although it has been noted that many of the preventive measures implemented to reduce SARS-CoV-2 incidence also have a clear benefit on reducing Mycobacterium tuberculosis (Mtb) transmission, 2020 saw the first year-over-year increase in TB deaths from 2005, regardless of physical distancing and PPE (personal protective equipment) wearing measures.[2,8]
In Canada, the pandemic significantly affected latent TB infection (LTBI) and active TB treatment, leading to ineffective measures for TB elimination.[9] In Spain,  newly diagnosed TB patients had more extended pulmonary disease, moreover there was a rise in household transmission probably due to anti-COVID-19 measures.[10] Also, in England, it has been observed a fall in rates of TB treatment initiation during the period of government-imposed lockdown (March 23–May 10, 2020), and an increase of cases of disseminated TB during the COVID-19 pandemic.[11] All this makes it important to evaluate the measures against COVID-19 globally and not only considering the pathologies related to SARS-CoV-2,
The COVID-19 emergence prompted the scientific community to focus on determining the mechanisms of transmission, the identification of virulence factors of SARS-CoV-2 and the development of suitable therapies.[12] Therapeutical management of COVID-19 is in constant change, and treatment guidelines are readily updated based on scientific evidence and experts’ opinion (National Institutes of Health, n.d.) as we entered in an era of “hype-based medicine”,[13] the long forgotten eminence-based medicine regained importance as the number of trials on possible therapies multiplied, some of them causing overnight changes in the management of COVID-19 patients.[14] The lack of antiviral therapies and the rapid spread of the infection convinced investigators and pharmaceutical companies to focus on the development of vaccines,[15] able  to induce neutralizing antibodies against SARS-CoV-2 Spike protein in naive subjects. The developed vaccines do not only trigger a humoral response against the protein, but they impact all the components of the immune response.
As most of the therapies used against COVID-19 disease therapies do not target SARS-CoV-2, but aim to regulate the host immune response[16,17] it is reasonable to consider the long-term effects of these therapies on subjects with latent TB infection (LTBI).
In this commentary, we aim to summarize treatments against SARS-CoV-2 and underline their possible effects on Mtb infection highlighting likely “side” effects that could help to contain virus-mediated damage and, conversely, prompt mycobacterial replication in both early infection or during Mtb latency.

Therapies Against SARS-CoV-2 Infection

SARS-CoV-2 represents the biggest therapeutic challenge of our century. At present, approximately 2900 clinical trials have been registered,[14] designing new molecules and repurposing existing drugs based on the virus biology and pathogenesis.
Therapeutical approaches range from convalescent plasma of people who have recovered from COVID-19, to medications which are commonly used to treat autoimmune or inflammatory diseases as well as drugs used to treat other infections.[18,19]
Pharmaceuticals used for COVID‑19 target different pathogenetic mechanisms, with the aim of a) blocking viral replication, summarized in points 1-3 of the Figure 1, and b) reducing tissue damage, modulating the immune responses, and preventing over-inflammation (Figure 1, points 4-6).


Figure 1 Figure 1. Schematic representation of the pharmaceuticals used against SARS-CoV-2 infection. The first class of molecules includes antivirals to prevent viral entry (point 1); the second class includes compounds that inhibit gene transcription (point 2) and the third class accounts molecules that prevent proteolytic processing and block viral docking (point 3). The points 4-6 described medications that reduce tissue damage, modulating the immune responses or preventing over-inflammation. 

The first class includes antivirals to prevent spike-protein-mediated cell fusion, thus blocking viral entry (Figure 1, point 1), inhibit gene transcription (Figure 1, point 2) or prevent proteolytic processing and block viral docking (Figure 1, point 3), as explained in Table 1.[20-23] Interestingly, several agents show no effects on SARS-CoV-2  even though they were described to have activity against other infections.[24]

Table 1.1 Table 1. Summary reporting experimental evidence of the impact of drugs used against SARS-CoV-2 infection on Mycobacterium tuberculosis infection.
Table 1.2

SARS-CoV-2 infection causes an overproduction of type I interferons triggering the transcription of several genes and the recruitment of CD4+ T helper lymphocytes, responsible for the Th1/Th2 response.[25] For this reason, immunomodulators (corticosteroids, interferons, monoclonal antibodies against inflammatory cytokines) have been suggested, and largely used, to reduce the over-inflammation that is responsible for several systemic disease manifestations.
However, the NHS Panel failed to evaluate the real role of some of these therapies due to insufficient evidence to recommend either for or against their use.[26]
Examples of drugs in this category are IL-1 inhibitors, colchicine, the antiparasitic agent ivermectin,[12] and thalidomide.[27] Some others are currently recommended as IL-6 inhibitors and Janus Kinase inhibitors (refer to Table 1 and Figure 1, point 4 to 6).
Although, the use of immunomodulatory treatments had an immediate impact on the care of patients infected with SARS-CoV-2, their long-term effects are unknown.

Impact of the Therapies Against SARS-CoV-2 on Mycobacterium tuberculosis

Mtb infection represents a classical model of persistent infection, a situation in which a microorganism can persist indefinitely within the host,[28,29] establishing an equilibrium between the pathogen and the host immune response whose modification could increase the risk of relapse and disease. Indeed, host immune response can limit Mtb spread, after macrophages killing evasion, creating a multicellular structure known as granuloma,[30] which entraps mycobacteria that persist in a heterogeneous range of states.[31] In the last decades, to deal with the emergence of Mtb strains resistant anti-TB drugs (MDR/RR-Mtb and XDR-Mtb), a novel approach has been proposed targeting the host and so named host directed therapies (HDTs).[32–35] HDTs can support antimycobacterial host response at different stages: a) perturbating granuloma integrity to enhance drug penetration; b) modifying autophagy or phagosome maturation to increase intracellular killing; c) promoting cell-mediated response; d) inducing antimicrobial peptides and controlling inflammation response by avoiding tissue damage.[36] While the use of HDTs seem to support anti-TB treatment in symptomatic individuals,[37] no data nor anecdotal knowledge support the use of such therapies in people with asymptomatic or subclinical infection.[38]
In other words, it is undeniable that some immunomodulatory treatments may alter the host-
Mtb equilibrium, favoring either the host or the bacteria. In this scenario, we cannot exclude that those immunomodulatory therapies used against COVID-19 may have a negative effect on infected individuals causing symptomatic TB.
A recent paper highlighted the relationship between SARS-CoV-2 and Mtb infection, showing that asymptomatic SARS-CoV-2 seropositive individuals with a positive IGRA exhibited heightened levels of humoral, cytokine production, and systemic inflammation compared to individuals negative for Mtb infection.[39
Mtb is apparently able to modulate the host immune response in SARS-CoV-2-infected individuals. Furthermore, various clinical cases describe TB reactivation following SARS-CoV-2 infection confirming the concerns that COVID-19 associated CD4+ T-cell depletion or altered T-cell function can have similar implications as HIV for TB disease progression, promoting the development of active TB.[25,40] Moreover, some studies highlighted a higher probability to develop severe disease in patients with SARS-CoV-2 / Mtb co-infection compared to COVID-19 patients.[41,42] Unfortunately, we have little information on TB occurrence after COVID-19 treatments.[43] On the other hand, there was a delay in the onset of the pandemics in many countries endemic for TB. Moreover, those countries showed lower COVID-19’s severe cases and SARS-CoV-2 related-mortality.[44] Intriguingly, one of the variables that was mathematically linked to COVID-19 low spread was BCG vaccination,[45] which is known to stimulate non-specific heterologous immune responses inducing cross-protective effects toward non-tuberculosis-related diseases, included SARS-CoV-2.[15,46,47] Indeed, numerous clinical trials are currently registered to update on the benefits of BCG vaccinations against SARS-CoV-2 exposure.[15,47]
We can classify therapies used against COVID-19 based on their activity on 
Mtb infection in four main drug classes: a) drugs acting directly on Mtb (Figure 2, point 1); b) drugs that modify phagosome acidification (Figure 2, point 2); c) drugs with adjuvant function that can indirectly modulate the infection (Figure 2, point 3) and d) drugs with immunomodulatory activity (Figure 2, point 4). Although many anti-COVID-19 pharmaceutics appeared to impair mycobacterial growth in in vitro experiments, (Figure 2, point 1),[48–51] we focus our attention on their immunomodulatory effects (Table 1). 

Figure 2 Figure 2. Schematic representation of the medications used against SARS-CoV-2 with effects on Mycobacterium tuberculosis infection. Therapies used against COVID-19 are classified based on their activity on Mtb infection in four main classes: drugs acting directly on mycobacteria (point 1) or indirectly showing ability to modify phagosome acidification (point 2), to modulate the infection with adjuvant functions (point 3) and to regulate hose immune response (point 4).

Hydroxychloroquine inactivates mycobacterial NAD+ dependent DNA ligase A[52] and modulates phagolysosome, reducing intracellular mycobacterial growth.[48] Similarly, chlorpromazine, an antipsychotic drug, has been observed to have antimycobacterial activity and to promote macrophagic killing by increasing phagosome acidification.[53] Acidification may though differently affect phagosomes at different stages of maturation.[54] Moreover, Mtb itself can influence phagosome maturation potentially counteracting these drugs.[55]
Nitazoxanide, which has been also suggested to modulate immune response inducing interferon-γ,[50,56] inhibits intracellular Mtb growth while amplifying Mtb-induced gene expression.
Thalidomide represents a compound that has been tested against Mtb infection showing a detrimental effect on infection control due to TNF-α inhibition with consequent increase in mycobacterial replication[57,58] (Figure 2 and Table 1).
Interestingly, several drugs that have been proposed against SARS-CoV-2 have not been tested in vitro against Mtb infection (Table 1). This is true for several molecules that act on the host immune system to prevent over inflammation (which has been observed as a critical point for the progression of SARS-CoV-2 infection). These compounds, that dampen pro-inflammatory cytokines, could impair the fine equilibrium between Mtb replication and the host immune system response, thus promoting active disease. Among them, monoclonal antibodies such as IL-6 antagonists and antivirals have been observed to significantly modulate host cytokine response and potentially alter host immune response versus Mtb replication.[59] Interestingly, Mtb regulates IL-6 secretion to inhibit type I interferon signaling and causes disease progression which appears to be associated to sigH gene expression.[60] For this reason, IL-6 antagonist could have important implications during 
Mtb infection.
Another example are corticosteroids that are beneficial in hospitalized COVID-19 patients,[61,62] but, conversely, could increase the risk of LTBI reactivation or progression of sub-clinical TB.


Conclusions

Given the specific effect of COVID-19 on T-cells and for anti-COVID-19 treatments on LTBI, clinicians should consider monitoring patients with both previous COVID-19 infection and LTBI to rapidly identify active disease and prevent Mtb transmission.


References   

  1. Furtado I, Aguiar A, Duarte R. Getting back on the road towards tuberculosis elimination: lessons learnt from the COVID-19 pandemic. J Bras Pneumol. 2021;47(2):e20210123. https://doi.org/10.36416/1806-3756/e20210123 PMid:33950099 PMCid:PMC8332847
  2. Pai M, Kasaeva T, Swaminathan S. Covid-19's Devastating Effect on Tuberculosis Care - A Path to Recovery. N Engl J Med [Internet]. 2022;0(0):null. https://doi.org/10.1056/NEJMp2118145 PMid:34986295
  3. Jain VK, Iyengar KP, Samy DA, Vaishya R. Tuberculosis in the era of COVID-19 in India. Diabetes Metab Syndr Clin Res Rev [Internet]. 2020;14(5):1439-43. https://doi.org/10.1016/j.dsx.2020.07.034 PMid:32755848 PMCid:PMC7387287
  4. Shaikh N, Pelzer PT, Thysen SM, Roy P, Harris RC, White RG. Impact of COVID-19 Disruptions on Global BCG Coverage and Paediatric TB Mortality: A Modelling Study. Vaccines. 2021 Oct;9(11). https://doi.org/10.3390/vaccines9111228 PMid:34835161 PMCid:PMC8624525
  5. Malik AA, Safdar N, Chandir S, Khan U, Khowaja S, Riaz N, et al. Tuberculosis control and care in the era of COVID-19. Health Policy Plan. 2020;35(8):1130-2. https://doi.org/10.1093/heapol/czaa109 PMid:32832996 PMCid:PMC7499582
  6. Janice K. Louie, Rocio Agraz-Lara LR, Felix Crespin, Lisa Chen 1, Graves S. Tuberculosis-Associated Hospitalizations and Deaths after COVID-19 ShelterIn-Place, San Francisco, California, USA. Emerg Infect Dis. 2021;27:2227-9. https://doi.org/10.3201/eid2708.210670 PMid:34287142 PMCid:PMC8314834
  7. Marwah V, Peter DK, Ajai Kumar T, Bhati G, Kumar A. Multidrug-resistant tuberculosis in COVID-19: Double trouble. Med J Armed Forces India [Internet]. 2021;77:S479-82. https://doi.org/10.1016/j.mjafi.2021.05.002 PMid:34334915 PMCid:PMC8313087
  8. Finn McQuaid C, McCreesh N, Read JM, Sumner T, Houben RMGJ, White RG, et al. The potential impact of COVID-19-related disruption on tuberculosis burden. Eur Respir J. 2020;56(2). https://doi.org/10.1183/13993003.01718-2020 PMid:32513784 PMCid:PMC7278504
  9. Geric C, Saroufim M, Landsman D, Richard J, Benedetti A, Batt J, et al. Impact of Covid-19 on Tuberculosis Prevention and Treatment in Canada: a multicentre analysis of 10,833 patients. J Infect Dis. 2021 Dec. https://doi.org/10.1093/infdis/jiab608 PMid:34919700 PMCid:PMC8755327
  10. Aznar ML, Espinosa-Pereiro J, Saborit N, Jové N, Sánchez Martinez F, Pérez-Recio S, et al. Impact of the COVID-19 pandemic on tuberculosis management in Spain. Int J Infect Dis IJID Off Publ Int Soc Infect Dis. 2021 Jul;108:300-5. https://doi.org/10.1016/j.ijid.2021.04.075 PMid:33930543 PMCid:PMC8078060
  11. Barrett J, Painter H, Rajgopal A, Keane D, John L, Papineni P, et al. Increase in disseminated TB during the COVID-19 pandemic. Vol. 25, The international journal of tuberculosis and lung disease : the official journal of the International Union against Tuberculosis and Lung Disease. France; 2021. p. 160-6. https://doi.org/10.5588/ijtld.20.0846 PMid:33656432
  12. Tu YF, Chien CS, Yarmishyn AA, Lin YY, Luo YH, Lin YT, et al. A review of sars-cov-2 and the ongoing clinical trials [Internet]. Vol. 21, International Journal of Molecular Sciences. 2020. p. 2657. https://doi.org/10.3390/ijms21072657 PMid:32290293 PMCid:PMC7177898
  13. Pearson H. How COVID broke the evidence pipeline. Nature. 2021;593(7858):182-5. https://doi.org/10.1038/d41586-021-01246-x PMid:33981057
  14. No authors listed. Evidence-based medicine: how COVID can drive positive change. Vol. 593, Nature. England; 2021. p. 168. https://doi.org/10.1038/d41586-021-01255-w PMid:33981058
  15. Gong W, Aspatwar A, Wang S, Parkkila S, Wu X. COVID-19 pandemic: SARS-CoV-2 specific vaccines and challenges, protection via BCG trained immunity, and clinical trials. Expert Rev Vaccines. 2021 Jul;20(7):857-80. https://doi.org/10.1080/14760584.2021.1938550 PMid:34078215 PMCid:PMC8220438
  16. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol [Internet]. 2021;19(3):141-54. https://doi.org/10.1038/s41579-020-00459-7 PMid:33024307 PMCid:PMC7537588
  17. Petersen E, Koopmans M, Go U, Hamer DH, Petrosillo N, Castelli F, et al. Personal View Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics. Lancet Infect Dis [Internet]. 2020;20(9):e238-44. https://doi.org/10.1016/S1473-3099(20)30484-9
  18. Nitulescu GM, Paunescu H, Moschos SA, Petrakis D, Nitulescu G, Ion GND, et al. Comprehensive analysis of drugs to treat SARS-CoV-2 infection: Mechanistic insights into current COVID-19 therapies (Review). Int J Mol Med. 2020;46(2):467-88. https://doi.org/10.3892/ijmm.2020.4608 PMid:32468014 PMCid:PMC7307820
  19. Joyner MJ, Wright RS, Fairweather DL, Senefeld JW, Bruno KA, Klassen SA, et al. Early safety indicators of COVID-19 convalescent plasma in 5,000 patients. medRxiv. 2020;130(9):4791-7. https://doi.org/10.1101/2020.05.12.20099879 PMid:32511566 PMCid:PMC7274247 
  20. Sarma P, Shekhar N, Prajapat M, Avti P, Kaur H, Kumar S, et al. In-silico homology assisted identification of inhibitor of RNA binding against 2019-nCoV N-protein (N terminal domain). J Biomol Struct Dyn. 2021 May;39(8):2724-2732. http://doi.org/10.1080/07391102.2020.1753580 PMID: 32266867
  21. O'keefe BR, Giomarelli B, Barnard DL, Shenoy SR, Chan PKS, Mcmahon JB, et al. Broad-Spectrum In Vitro Activity and In Vivo Efficacy of the Antiviral Protein Griffithsin against Emerging Viruses of the Family Coronaviridae. J Virol [Internet]. 2010;84(5):2511-21. https://doi.org/10.1128/JVI.02322-09 PMid:20032190 PMCid:PMC2820936
  22. Plaze M, Attali D, Petit AC, Blatzer M, Simon-Loriere E, Vinckier F, et al. Repurposing chlorpromazine to treat COVID-19: The reCoVery study. Encephale. 2020 Jun 1;46(3):169-72. https://doi.org/10.1016/j.encep.2020.05.006 PMid:32425222 PMCid:PMC7229964
  23. Infante M, Ricordi C, Alejandro R, Caprio M, Fabbri A. Hydroxychloroquine in the COVID-19 pandemic era: in pursuit of a rational use for prophylaxis of SARS-CoV-2 infection. Expert Rev Anti Infect Ther. 2020; https://doi.org/10.1080/14787210.2020.1799785 PMid:32693652 PMCid:PMC7441799
  24. Mina T. Kellen. Letter to the Editor Nitazoxanide/azithromycin combination for COVID-19: A suggested new protocol for early management. Pharmacol Res J [Internet]. 2020; https://doi.org/10.20944/preprints202004.0432.v1
  25. Riou C, du Bruyn E, Stek C, Daroowala R, Goliath RT, Abrahams F, et al. Relationship of SARS-CoV-2-specific CD4 response to COVID-19 severity and impact of HIV-1 and tuberculosis coinfection. J Clin Invest [Internet]. 2021;131(12). https://doi.org/10.1172/JCI149125 PMid:33945513 PMCid:PMC8203446
  26. National Institutes of Health. COVID-19 Treatment Guidelines Panel. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines [Internet]. [cited 2021 Aug 12].
  27. Chen C, Qi F, Shi K, Li Y, Li J, Chen Y, et al. Thalidomide Combined with Low-dose Glucocorticoid in the Treatment of COVID-19 Pneumonia [Internet]. Preprints; 2020 [cited 2021 Aug 13].
  28. Kane M, Golovkina T. Common Threads in Persistent Viral Infections. J Virol. 2010;84(9):4116-23. https://doi.org/10.1128/JVI.01905-09 PMid:19955304 PMCid:PMC2863747
  29. Fisher RA, Gollan B, Helaine S. Persistent bacterial infections and persister cells. Nat Rev Microbiol. 2017 Aug;15(8):453-64. https://doi.org/10.1038/nrmicro.2017.42 PMid:28529326
  30. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat Immunol. 2009;10(9):943-8. https://doi.org/10.1038/ni.1781 PMid:19692995 PMCid:PMC2759071
  31. Delogu G, Sali M, Fadda G. The biology of mycobacterium tuberculosis infection. Mediterr J Hematol Infect Dis. 2013;5(1). https://doi.org/10.4084/mjhid.2013.070 PMid:24363885 PMCid:PMC3867229
  32. Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discov [Internet]. 2018;17(1):35-56. https://doi.org/10.1038/nrd.2017.162 PMid:28935918 PMCid:PMC7097079
  33. Palucci I, Maulucci G, De Maio F, Sali M, Romagnoli A, Petrone L, et al. Inhibition of Transglutaminase 2 as a Potential Host-Directed Therapy Against Mycobacterium tuberculosis. Front Immunol. 2020;10(January):1-13. https://doi.org/10.3389/fimmu.2019.03042 PMid:32038614 PMCid:PMC6992558
  34. Kumar R, Kolloli A, Singh P, Vinnard C, Kaplan G, Subbian S. Thalidomide and Phosphodiesterase 4 Inhibitors as Host Directed Therapeutics for Tuberculous Meningitis: Insights From the Rabbit Model. Front Cell Infect Microbiol. 2019;9:450. https://doi.org/10.3389/fcimb.2019.00450 PMid:32010638 PMCid:PMC6972508
  35. Costa DL, Maiga M, Subbian S. Editorial: Host-Directed Therapies for Tuberculosis. Front Cell Infect Microbiol [Internet]. 2021;11:736. https://doi.org/10.3389/fcimb.2021.742053 PMid:34422685 PMCid:PMC8377667
  36. Kolloli A, Kumar R, Singh P, Narang A, Kaplan G, Sigal A, et al. Aggregation state of Mycobacterium tuberculosis impacts host immunity and augments pulmonary disease pathology. Commun Biol. 2021 Nov;4(1):1256. https://doi.org/10.1038/s42003-021-02769-9 PMid:34732811 PMCid:PMC8566596
  37. Wallis RS, Ginindza S, Beattie T, Arjun N, Likoti M, Edward VA, et al. Adjunctive host-directed therapies for pulmonary tuberculosis: a prospective, open-label, phase 2, randomised controlled trial. Lancet Respir Med [Internet]. 2021;9(8):897-908. https://doi.org/10.1016/S2213-2600(20)30448-3
  38. Richards AS, Sossen B, Emery JC, Horton KC, Heinsohn T, Frascella B, et al. The natural history of TB disease-a synthesis of data to quantify progression and regression across the spectrum. medRxiv [Internet]. 2021; https://doi.org/10.1101/2021.09.13.21263499
  39. Rajamanickam A, Kumar NP, Padmapriyadarsini C, Nancy A, Selvaraj N, Karunanithi K, et al. Latent tuberculosis co-infection is associated with heightened levels of humoral, cytokine and acute phase responses in seropositive SARS-CoV-2 infection. J Infect [Internet]. 2021;83(3):339-46. https://doi.org/10.1016/j.jinf.2021.07.029 PMid:34329676 PMCid:PMC8316716
  40. Khayat M, Fan H, Vali Y. COVID-19 promoting the development of active tuberculosis in a patient with latent tuberculosis infection: A case report. Respir Med Case Reports. 2021 Jan 1;32:101344. https://doi.org/10.1016/j.rmcr.2021.101344 PMid:33495728 PMCid:PMC7816563
  41. Mousquer GT, Peres A, Fiegenbaum M. Pathology of TB/COVID-19 Co-Infection: The phantom menace. Tuberculosis (Edinb). 2021 Jan;126:102020. https://doi.org/10.1016/j.tube.2020.102020 PMid:33246269 PMCid:PMC7669479
  42. Gao Y, Liu M, Chen Y, Shi S, Geng J, Tian J. Association between tuberculosis and COVID-19 severity and mortality: A rapid systematic review and meta-analysis. Vol. 93, Journal of medical virology. 2021. p. 194-6. https://doi.org/10.1002/jmv.26311 PMid:32687228 PMCid:PMC7405273
  43. Tadolini M, Codecasa LR, García-García JM, Blanc FX, Borisov S, Alffenaar JW, et al. Active tuberculosis, sequelae and COVID-19 co-infection: First cohort of 49 cases. Eur Respir J. 2020;56(1). https://doi.org/10.1183/13993003.02328-2020 PMid:32586888 PMCid:PMC7315815
  44. Lalaoui R, Bakour S, Raoult D, Verger P, Sokhna C, Devaux C, et al. What could explain the late emergence of COVID-19 in Africa? New microbes new Infect. 2020 Nov;38:100760. https://doi.org/10.1016/j.nmni.2020.100760 PMid:32983542 PMCid:PMC7508045
  45. Zawbaa HM, El-Gendy A, Saeed H, Osama H, Ali AMA, Gomaa D, et al. A study of the possible factors affecting COVID-19 spread, severity and mortality and the effect of social distancing on these factors: Machine learning forecasting model. Int J Clin Pract. 2021 Jun;75(6):e14116. https://doi.org/10.1111/ijcp.14116 PMCid:PMC7995223
  46. Covián C, Retamal-Díaz A, Bueno SM, Kalergis AM. Could BCG Vaccination Induce Protective Trained Immunity for SARS-CoV-2? Front Immunol [Internet]. 2020;11. https://doi.org/10.3389/fimmu.2020.00970 PMid:32574258 PMCid:PMC7227382
  47. Giamarellos-Bourboulis EJ, Tsilika M, Moorlag S, Antonakos N, Kotsaki A, Domínguez-Andrés J, et al. Activate: Randomized Clinical Trial of BCG Vaccination against Infection in the Elderly. Cell. 2020 Oct;183(2):315-323.e9. https://doi.org/10.1016/j.cell.2020.08.051 PMid:32941801 PMCid:PMC7462457
  48. Rolain JM, Colson P, Raoult D. Recycling of chloroquine and its hydroxyl analogue to face bacterial, fungal and viral infections in the 21st century. Int J Antimicrob Agents. 2007;30(4):297-308. https://doi.org/10.1016/j.ijantimicag.2007.05.015 PMid:17629679 PMCid:PMC7126847
  49. Lim LE, Vilchèze C, Ng C, Jacobs WR, Ramón-García S, Thompson CJ. Anthelmintic avermectins kill mycobacterium tuberculosis, including multidrug-resistant clinical strains. Antimicrob Agents Chemother. 2013;57(2):1040-6. https://doi.org/10.1128/AAC.01696-12 PMid:23165468 PMCid:PMC3553693
  50. Ranjbar S, Haridas V, Nambu A, Jasenosky LD, Sadhukhan S, Ebert TS, et al. Cytoplasmic RNA Sensor Pathways and Nitazoxanide Broadly Inhibit Intracellular Mycobacterium tuberculosis Growth. iScience [Internet]. 2019;22:299-313. https://doi.org/10.1016/j.isci.2019.11.001 PMid:31805434 PMCid:PMC6909047
  51. Sriram D, Yogeeswari P, Dinakaran M, Sowmya M. Synthesis, anti-HIV and antitubercular activities of nelfinavir diester derivatives. Biomed Pharmacother [Internet]. 2008;62(1):1-5. https://doi.org/10.1016/j.biopha.2007.08.002 PMid:17890044
  52. Singh V, Somvanshi P. Toward the Virtual Screening of Potential Drugs in the Homology Modeled NAD+ Dependent DNA Ligase from Mycobacterium tuberculosis. Protein Pept Lett. 2010;17(2):269-76. https://doi.org/10.2174/092986610790225950 PMid:20214650
  53. Machado D, Pires D, Couto I. Ion Channel Blockers as Antimicrobial Agents , Efflux Inhibitors , and Enhancers of Macrophage Killing Activity against Drug Resistant Mycobacterium tuberculosis. 2016;1-28. https://doi.org/10.1371/journal.pone.0149326 PMid:26919135 PMCid:PMC4769142
  54. Romagnoli A, Petruccioli E, Palucci I, Camassa S, Carata E, Petrone L, et al. Clinical isolates of the modern Mycobacterium tuberculosis lineage 4 evade host defense in human macrophages through eluding IL-1β-induced autophagy article. Cell Death Dis [Internet]. 2018;9(6). https://doi.org/10.1038/s41419-018-0640-8 PMid:29795378 PMCid:PMC5967325
  55. Ramachandra L, Smialek JL, Shank SS, Convery M, Boom WH, Harding C V. Phagosomal processing of Mycobacterium tuberculosis antigen 85B is modulated independently of mycobacterial viability and phagosome maturation. Infect Immun. 2005 Feb;73(2):1097-105. https://doi.org/10.1128/IAI.73.2.1097-1105.2005 PMid:15664953 PMCid:PMC547092
  56. Iacobino A, Giannoni F, Pardini M, Piccaro G. The Combination Rifampin-Nitazoxanide, but Not Rifampin-Isoniazid-Pyrazinamide-Ethambutol, Kills Dormant Mycobacterium tuberculosis in Hypoxia at Neutral pH. Antimicrob Agents Chemother. 2019;(April):1-4. https://doi.org/10.1128/AAC.00273-19 PMid:31010861 PMCid:PMC6591638
  57. Verbon A, Juffermans NP, Speelman P, Van Deventer SJH, Ten Berge IJM, Guchelaar HJ, et al. A single oral dose of thalidomide enhances the capacity of lymphocytes to secrete gamma interferon in healthy humans. Antimicrob Agents Chemother. 2000;44(9):2286-90. https://doi.org/10.1128/AAC.44.9.2286-2290.2000 PMid:10952569 PMCid:PMC90059
  58. Wang L, Hong Y, Wu J, Leung YK, Huang Y. Efficacy of thalidomide therapy in pediatric Crohn's disease with evidence of tuberculosis. World J Gastroenterol. 2017;23(43):7727-34. https://doi.org/10.3748/wjg.v23.i43.7727 PMid:29209113 PMCid:PMC5703932
  59. Urdahl KB, Shafiani S, Ernst JD. Initiation and regulation of T-cell responses in tuberculosis. Vol. 4, Mucosal Immunology. 2011. p. 288-93. https://doi.org/10.1038/mi.2011.10 PMid:21451503 PMCid:PMC3206635
  60. Martinez AN, Mehra S, Kaushal D. Role of interleukin 6 in innate immunity to Mycobacterium tuberculosis infection. J Infect Dis. 2013 Apr;207(8):1253-61. https://doi.org/10.1093/infdis/jit037 PMid:23359591 PMCid:PMC3693587
  61. The RECOVERY Collaborative Group. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med [Internet]. 2020 Jul 17;384(8):693-704. https://doi.org/10.1056/NEJMoa2021436 PMid:32678530 PMCid:PMC7383595
  62. Liu J, Zhang S, Dong X, Li Z, Xu Q, Feng H, et al. Corticosteroid treatment in severe COVID-19 patients with acute respiratory distress syndrome. J Clin Invest. 2020 Dec;130(12):6417-28. https://doi.org/10.1172/JCI140617 PMid:33141117 PMCid:PMC7685724

[TOP]